Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
1.
J Complement Integr Med ; 19(3): 661-668, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35106983

ABSTRACT

OBJECTIVES: Anemia is a direct or indirect consequence of oxidative stress via free radicals on erythrocytes and subsequently on other tissues like liver. Ficus glumosa constitute a rich pharmacologically compound that can prevent or repair oxidative damage. Therefore, this study seeks to evaluate the effect of F. glumosa on phenylhydrazine-induced hemolytic anemia and hepatic damage in rats. METHODS: Twenty-four (24) albino Wistar rats were assigned to four (4) experimental groups (n=6) as follows: Group I (non-anemic control) and Group 2 (anemic control) received normal saline, while Group III and IV (test groups) 200 and 400 mg/kg of aqueous leaf extract of F. glumosa (ALEFG), respectively. All the groups were treated orally (via a cannula) for seven consecutive days. Intraperitoneal (IP) injection of phenylhydrazine (PHZ) at 40 mg/kg for two consecutive days induced hemolytic anemia in group II to IV before treatment. Rats of all groups were anaesthetized and sacrificed 24 h after the last treatment. Blood and liver samples were collected for some hematological indices, liver function test, antioxidant parameter and histological analysis. RESULTS: The LD50 of ALEFG was assessed orally in rats and found to be above 5,000 mg/kg body weight. Significant (p<0.05) decreases in the level of red blood cell (RBC), hemoglobin (HGB) concentrations and packed cell volume (PCV) by 50% after 2 days of PHZ induction, were attenuated by more than 50% after 7 days administration of ALEFG at 200 and 400 mg/kg. The percentage change in body weight increased significantly (p<0.05) after 7 days post PHZ-induced anemia, but those that received oral administration of ALEFG (at 200 and 400 mg/kg) for 7 days increased significantly (p<0.05) by more than 2%, dose-dependently compared to anemic untreated group. Increased level of serum ALT, AST, ALP and GGT in PHZ-induced anemic animals, were significantly (p<0.05) attenuated in the groups that received oral administration of ALEFG (at 200 and 400 mg/kg) for 7 days. Decreased level of catalase (CAT) and superoxide dismutase (SOD) activities with concomitant increase in malondialdehyde (MDA) content from PHZ-induced untreated group, were significantly (p<0.05) mitigated in the rats that received oral administration of ALEFG (at 200 and 400 mg/kg) for 7 days. Histopathological analysis showed that ALEFG could remarkably though not completely mitigated PHZ-induced hepatic damage. CONCLUSIONS: Our data suggests that the leaves of F. glumosa contain important antioxidant(s) that could effectively reduce hemolytic anemia and hepatic damage, especially during phenylhydrazine-induced toxicity.


Subject(s)
Anemia, Hemolytic , Ficus , Anemia, Hemolytic/chemically induced , Anemia, Hemolytic/drug therapy , Anemia, Hemolytic/prevention & control , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Body Weight , Catalase , Hemoglobins , Malondialdehyde , Oxidative Stress , Phenylhydrazines/adverse effects , Plant Extracts/adverse effects , Rats , Rats, Wistar , Saline Solution/adverse effects , Superoxide Dismutase
2.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34507997

ABSTRACT

Late-stage anthrax infections are characterized by dysregulated immune responses and hematogenous spread of Bacillus anthracis, leading to extreme bacteremia, sepsis, multiple organ failure, and, ultimately, death. Despite the bacterium being nonhemolytic, some fulminant anthrax patients develop a secondary atypical hemolytic uremic syndrome (aHUS) through unknown mechanisms. We recapitulated the pathology in baboons challenged with cell wall peptidoglycan (PGN), a polymeric, pathogen-associated molecular pattern responsible for the hemostatic dysregulation in anthrax sepsis. Similar to aHUS anthrax patients, PGN induces an initial hematocrit elevation followed by progressive hemolytic anemia and associated renal failure. Etiologically, PGN induces erythrolysis through direct excessive activation of all three complement pathways. Blunting terminal complement activation with a C5 neutralizing peptide prevented the progressive deposition of membrane attack complexes on red blood cells (RBC) and subsequent intravascular hemolysis, heme cytotoxicity, and acute kidney injury. Importantly, C5 neutralization did not prevent immune recognition of PGN and shifted the systemic inflammatory responses, consistent with improved survival in sepsis. Whereas PGN-induced hemostatic dysregulation was unchanged, C5 inhibition augmented fibrinolysis and improved the thromboischemic resolution. Overall, our study identifies PGN-driven complement activation as the pathologic mechanism underlying hemolytic anemia in anthrax and likely other gram-positive infections in which PGN is abundantly represented. Neutralization of terminal complement reactions reduces the hemolytic uremic pathology induced by PGN and could alleviate heme cytotoxicity and its associated kidney failure in gram-positive infections.


Subject(s)
Acute Kidney Injury/prevention & control , Anemia, Hemolytic/prevention & control , Bacillus anthracis/chemistry , Cell Wall/chemistry , Complement C5/antagonists & inhibitors , Peptidoglycan/toxicity , Sepsis/complications , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Anemia, Hemolytic/etiology , Anemia, Hemolytic/pathology , Animals , Anthrax/microbiology , Anthrax/pathology , Female , Hemolysis , Male , Papio , Sepsis/chemically induced
4.
Article in Spanish | LILACS | ID: biblio-1388633

ABSTRACT

RESUMEN La enfermedad hemolítica perinatal es infrecuente hoy por la prevención que de ella se hace. Sin embargo, existen casos de madres altamente sensibilizadas que desean tener un hijo, lo que obliga a que ese embarazo deseado sea controlado de manera especial y sometido a procedimientos invasivos no exentos de morbimortalidad fetal. El uso prenatal de inmunoglobulina humana en la madre puede representar una alternativa terapéutica. Se presenta un caso en que su uso impidió el desarrollo de enfermedad intrauterina y favoreció la buena evolución neonatal a pesar de que el pronóstico inicial era muy adverso.


ABSTRACT Perinatal Hemolytic Disease is uncommon today due to its prevention. However, there are cases of highly sensitized mothers who wish to have a child, that forces this desired pregnancy to be controlled in a special way and be subjected to invasive procedures not exempt from fetal morbidity and mortality. Prenatal use of human inmunoglobulin in the mother may represent a therapeutic alternative. We present a case in which its use prevented the development of intrauterine disease and favored a good neonatal evolution despite the fact that the initial prognosis was very adverse.


Subject(s)
Humans , Female , Pregnancy , Infant, Newborn , Adult , Immunoglobulins, Intravenous/administration & dosage , Erythroblastosis, Fetal/prevention & control , Anemia, Hemolytic/prevention & control , Prenatal Care , Rh Isoimmunization/prevention & control , Blood Transfusion, Intrauterine
5.
Transfusion ; 60(7): 1381-1390, 2020 07.
Article in English | MEDLINE | ID: mdl-32488887

ABSTRACT

BACKGROUND: Intravenous immunoglobulins (IVIG) are derived from large human plasma pools. IVIG-associated hemolytic anemia (HA) is a known class effect, likely attributed to dose-dependent passive transfer of anti-A/B isoagglutinins. Two isoagglutinin reduction steps were implemented in the manufacturing process of Privigen (human 10% liquid IVIG): exclusion of high-anti-A-titer donors in 2013, replaced by specific immunoaffinity chromatography in 2015. We aim to estimate the clinical effectiveness of both measures. STUDY DESIGN AND METHODS: Using the US hospital-based Premier Healthcare Database, three Privigen cohorts were generated based on calendar periods indicative of manufacturing changes: Period 1 (baseline) January 2008 to December 2012, Period 2 (high-anti-A-titer donor exclusion) October 2013 to December 2015, and Period 3 (immunoaffinity chromatography) October 2016 to April 2019. HA within a 10-day at-risk period after Privigen administrations was identified from review of patient record summaries. Incidence rate ratios (IRRs) were estimated from Poisson regression (Period 1 reference) adjusting for hospital setting, sex, age, Privigen indication, dose, and first use. RESULTS: Crude incidence rates of HA were 1.49 per 10,000 person-days in Period 1 (38 HA, 9439 patients), 1.01 in Period 2 (20 HA, 7710 patients), and 0.14 in Period 3 (3 HA, 7759 patients). Adjusted IRR for HA in Period 2 was 0.71 (95% confidence interval [CI], 0.41-1.23), and in Period 3 was 0.10 (0.03-0.33) compared with Period 1. The IRR for HA in Period 3 compared with Period 2 was 0.14 (95% CI, 0.04-0.47). CONCLUSION: Implementation of immunoaffinity chromatography in Privigen manufacturing resulted in a significant 90% reduction of HA risk. HA has become a rare event in association with Privigen use.


Subject(s)
ABO Blood-Group System , Anemia, Hemolytic , Hemagglutinins , Immunoglobulins, Intravenous , Adult , Aged , Anemia, Hemolytic/chemically induced , Anemia, Hemolytic/epidemiology , Anemia, Hemolytic/prevention & control , Chromatography, Affinity , Female , Hemagglutinins/administration & dosage , Hemagglutinins/adverse effects , Hemagglutinins/chemistry , Humans , Immunoglobulins, Intravenous/administration & dosage , Immunoglobulins, Intravenous/adverse effects , Immunoglobulins, Intravenous/chemistry , Incidence , Male , Middle Aged , Retrospective Studies
7.
Malar J ; 19(1): 39, 2020 Jan 22.
Article in English | MEDLINE | ID: mdl-31969146

ABSTRACT

BACKGROUND: Parenteral artesunate is the treatment of choice for severe malaria. It is safe, efficacious and well tolerated anti-malarial. However, delayed haemolysis has been reported in travellers, non-immune individuals and in African children. METHODS: A prospective, observational study was carried out in admitted severe malaria patients receiving parenteral artesunate. The patients were followed up until day 28 for monitoring clinical as well as laboratory parameters for haemolytic anaemia. RESULTS: Twenty-four patients with severe malaria receiving injection artesunate were enrolled in the study. Post-artesunate delayed haemolysis following parenteral artesunate therapy was observed in three of 24 patients (12.5%, 95% confidence interval 4.5-31.2%). Haemolysis was observed in two more patients possibly due to other reasons. The haemoglobin fall ranged from 13.6 to 38.3% from day 7 to day 28 in these patients. CONCLUSION: The possibility of delayed haemolysis should be considered while treating the severe malaria patients with parenteral artesunate. The study highlights the need for further studies in different epidemiological settings.


Subject(s)
Anemia, Hemolytic/prevention & control , Antimalarials/administration & dosage , Artesunate/administration & dosage , Malaria/drug therapy , Administration, Intravenous , Adolescent , Adult , Anemia, Hemolytic/chemically induced , Child , Child, Preschool , Female , Hemolysis/drug effects , Humans , India , Infant , Malaria/blood , Male , Middle Aged , Prospective Studies , Time Factors , Young Adult
8.
Am J Hematol ; 95(11): 1334-1343, 2020 11.
Article in English | MEDLINE | ID: mdl-33464651

ABSTRACT

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired, life-threatening hematologic disease characterized by chronic complement-mediated hemolysis and thrombosis. Despite treatment with eculizumab, a C5 inhibitor, 72% of individuals remain anemic. Pegcetacoplan (APL-2), a PEGylated C3 inhibitor, has the potential to provide more complete hemolysis control in patients with PNH. This open-label, phase Ib study was designed to assess the safety, tolerability, and pharmacokinetics of pegcetacoplan in subjects with PNH who remained anemic during treatment with eculizumab. Pharmacodynamic endpoints were also assessed as an exploratory objective of this study. Data are presented for six subjects in cohort 4 who received treatment for up to 2 years. In total, 427 treatment-emergent adverse events (TEAEs) were reported, 68 of which were possibly related to the study drug. Eight serious TEAEs occurred in two subjects; three of these events were considered possibly related to the study drug. Pegcetacoplan pharmacokinetic concentrations accumulated with repeated dosing, and steady state was reached at approximately 6-8 weeks. Lactate dehydrogenase levels were well controlled by eculizumab at baseline. Pegcetacoplan increased hemoglobin levels and decreased both reticulocyte count and total bilirubin in all six subjects. Improvements were observed in Functional Assessment of Chronic Illness Therapy Fatigue scores. Two subjects discontinued for reasons unrelated to pegcetacoplan. All four subjects who completed the study transitioned to pegcetacoplan monotherapy following eculizumab discontinuation and avoided transfusions. In this small study, pegcetacoplan therapy was generally well-tolerated, and resulted in an improved hematological response by achieving broad hemolysis control, enabling eculizumab discontinuation.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Complement C3/antagonists & inhibitors , Hemoglobinuria, Paroxysmal/drug therapy , Adult , Anemia, Hemolytic/drug therapy , Anemia, Hemolytic/etiology , Anemia, Hemolytic/prevention & control , Antibodies, Monoclonal, Humanized/adverse effects , Bilirubin/blood , Chemical and Drug Induced Liver Injury/etiology , Complement C5/antagonists & inhibitors , Drug Substitution , Female , Fever/chemically induced , Hemoglobins/analysis , Hemoglobinuria, Paroxysmal/blood , Hemoglobinuria, Paroxysmal/immunology , Hemolysis/drug effects , Humans , L-Lactate Dehydrogenase/blood , Male , Middle Aged , Pancreatitis/chemically induced , Prospective Studies , Reticulocyte Count
9.
Transfus Clin Biol ; 26(2): 99-101, 2019 May.
Article in English | MEDLINE | ID: mdl-30926306

ABSTRACT

Post-transfusion hemolysis is the most frequent immune reaction to transfusion in sickle cell disease. Its frequency is underestimated due to its biological and clinical characteristics. It results principally from the high incidence of alloimmunization in these patients, but no antibodies are detectable in 30% of cases. Prevention is based on the prevention of alloimmunization through the use of matched RBCs for highly immunogenic blood groups, taking into account the patient's transfusion history, particularly in patients undergoing occasional transfusion, which is associated with a higher risk of DHTR development than chronic transfusion. In addition to the use of matched RBCs, the prevention of alloimmunization through immunotherapy should be considered.


Subject(s)
Anemia, Hemolytic/prevention & control , Anemia, Sickle Cell/complications , Transfusion Reaction/prevention & control , Anemia, Hemolytic/etiology , Anemia, Hemolytic/immunology , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/therapy , Autoantibodies/biosynthesis , Autoantibodies/immunology , Blood Group Incompatibility/complications , Hemolysis , Humans , Immunosuppressive Agents/therapeutic use , Isoantibodies/biosynthesis , Isoantibodies/immunology , Premedication , Risk Assessment , Rituximab/therapeutic use , Time Factors , Transfusion Reaction/immunology
10.
Interact Cardiovasc Thorac Surg ; 28(6): 994-995, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30715380

ABSTRACT

Paroxysmal nocturnal haemoglobinuria is a rare disorder characterized by haemolytic anaemia and pancytopaenia. The use of cardiopulmonary bypass can lead to a haemolytic crisis in patients with paroxysmal nocturnal haemoglobinuria due to activation of complement-mediated haemolysis. We report the successful management of a 69-year-old man undergoing aortic valve replacement with standard heparin-protamine protocol by using eculizumab, a monoclonal antibody of complement factor C5. The surgery was performed without triggering a haemolytic crisis, and the patient was discharged from the hospital without major complications.


Subject(s)
Anemia, Hemolytic/prevention & control , Antibodies, Monoclonal, Humanized/therapeutic use , Aortic Valve Stenosis/surgery , Aortic Valve/surgery , Heart Valve Prosthesis Implantation/methods , Hemoglobinuria, Paroxysmal/drug therapy , Postoperative Complications/prevention & control , Aged , Anemia, Hemolytic/etiology , Aortic Valve Stenosis/complications , Hemoglobinuria, Paroxysmal/complications , Humans , Male , Postoperative Complications/etiology
11.
Biol Trace Elem Res ; 189(2): 490-500, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30151563

ABSTRACT

Selenium (Se), an essential trace element and potent nutritional antioxidant, exerts its biological effects through incorporation into selenoproteins like glutathione peroxidase (GPx). Modest decrement in the levels of GPx could be partly responsible for peroxidation of RBCs, which results into hemolytic anemia. Therefore, it is hypothesized that dietary Se, as selenoproteins (GPx), can maintain the homeostasis in RBCs and regulate the erythropoiesis by preventing oxidative stress-mediated hemolysis. Se-deficient (0.01 ppm), Se-adequate (0.1 ppm sodium selenite), and Se-supplemented (0.5 ppm sodium selenite) status were created in Balb/c mice by feeding yeast-based diets for 8 weeks and established by measuring Se levels in plasma and activities, expressions of Se-dependent selenoproteins. Fifty percent of mice from each differential Se group were treated with phenylhydrazine (PHZ, 20 mg/kg, i.p.) to induce hemolytic anemia. Results indicated that PHZ-treated Se-deficient animals demonstrated increased hemolysis, abnormal RBC morphology, increase in Heinz bodies and reticulocytes, and denaturation of hemoglobin to globin precipitates and methemoglobin. Se supplementation protected against these hemolytic changes and makes RBCs less fragile. These findings were consistent with dietary Se concentration-dependent changes in activity and expression of GPx indicating that ROS-mediated oxidative stress is integral to hemolysis. Protective effects of Se supplementation against increased levels of ROS, protein carbonyls, and peroxide damage to membrane lipids and enzymatic antioxidants validated these observations. In conclusion, dietary Se supplementation protected the RBCs against hemolysis by mitigating ROS-mediated oxidative stress.


Subject(s)
Anemia, Hemolytic/metabolism , Anemia, Hemolytic/prevention & control , Selenium/therapeutic use , Anemia, Hemolytic/chemically induced , Animals , Antioxidants/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Glutathione Peroxidase/metabolism , Hemolysis/drug effects , Homeostasis/drug effects , Mice , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Phenylhydrazines/toxicity , Reactive Oxygen Species/metabolism , Sodium Selenite/therapeutic use
12.
Methods Mol Biol ; 1885: 347-359, 2019.
Article in English | MEDLINE | ID: mdl-30506209

ABSTRACT

RhD negative pregnant women who carry an RhD positive fetus are at risk of immunization against the D antigen, which may result in hemolytic disease of the fetus and the newborn. Predicting the fetal RhD status by noninvasive antenatal screening for the fetal RhD gene (RHD) can guide targeted use of antenatal anti-D prophylaxis.Cell-free fetal DNA is extracted from maternal plasma from RhD negative pregnant women at a gestational age of 25 weeks. A real-time PCR-based detection of two RHD exons enables reliable prediction of the fetal RhD status to determine the administration of antenatal prophylaxis, as well as postnatal prophylaxis.


Subject(s)
Anemia, Hemolytic/etiology , Anemia, Hemolytic/prevention & control , Blood Grouping and Crossmatching/methods , Prenatal Diagnosis/methods , Rh-Hr Blood-Group System , Rho(D) Immune Globulin , Female , Humans , Pregnancy , Real-Time Polymerase Chain Reaction , Rh-Hr Blood-Group System/genetics , Rh-Hr Blood-Group System/immunology , Rho(D) Immune Globulin/genetics , Rho(D) Immune Globulin/immunology
13.
Blood ; 133(9): 893-901, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30559259

ABSTRACT

Cold agglutinin disease is a difficult-to-treat autoimmune hemolytic anemia in which immunoglobulin M antibodies bind to erythrocytes and fix complement, resulting in predominantly extravascular hemolysis. This trial tested the hypothesis that the anti-C1s antibody sutimlimab would ameliorate hemolytic anemia. Ten patients with cold agglutinin disease participated in the phase 1b component of a first-in-human trial. Patients received a test dose of 10-mg/kg sutimlimab followed by a full dose of 60 mg/kg 1 to 4 days later and 3 additional weekly doses of 60 mg/kg. All infusions were well tolerated without premedication. No drug-related serious adverse events were observed. Seven of 10 patients with cold agglutinin disease responded with a hemoglobin increase >2 g/dL. Sutimlimab rapidly increased hemoglobin levels by a median of 1.6 g/dL within the first week, and by a median of 3.9 g/dL (interquartile range, 1.3-4.5 g/dL; 95% confidence interval, 2.1-4.5) within 6 weeks (P = .005). Sutimlimab rapidly abrogated extravascular hemolysis, normalizing bilirubin levels within 24 hours in most patients and normalizing haptoglobin levels in 4 patients within 1 week. Hemolytic anemia recurred when drug levels were cleared from the circulation 3 to 4 weeks after the last dose of sutimlimab. Reexposure to sutimlimab in a named patient program recapitulated the control of hemolytic anemia. All 6 previously transfused patients became transfusion-free during treatment. Sutimlimab was safe, well tolerated, and rapidly stopped C1s complement-mediated hemolysis in patients with cold agglutinin disease, significantly increasing hemoglobin levels and precluding the need for transfusions. This trial was registered at www.clinicaltrials.gov as #NCT02502903.


Subject(s)
Anemia, Hemolytic, Autoimmune/drug therapy , Anemia, Hemolytic/prevention & control , Antibodies, Monoclonal, Humanized/therapeutic use , Complement C1s/antagonists & inhibitors , Hemolysis/drug effects , Severity of Illness Index , Aged , Anemia, Hemolytic/etiology , Anemia, Hemolytic, Autoimmune/complications , Complement C1s/immunology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Prospective Studies
14.
Am J Obstet Gynecol ; 219(3): 291.e1-291.e9, 2018 09.
Article in English | MEDLINE | ID: mdl-29902448

ABSTRACT

BACKGROUND: Intrauterine transfusion for severe alloimmunization in pregnancy performed <20 weeks' gestation is associated with a higher fetal death rate. Intravenous immunoglobulins may prevent hemolysis and could therefore be a noninvasive alternative for early transfusions. OBJECTIVE: We evaluated whether maternal treatment with intravenous immunoglobulins defers the development of severe fetal anemia and its consequences in a retrospective cohort to which 12 fetal therapy centers contributed. STUDY DESIGN: We included consecutive pregnancies of alloimmunized women with a history of severe hemolytic disease and by propensity analysis compared index pregnancies treated with intravenous immunoglobulins (n = 24) with pregnancies managed without intravenous immunoglobulins (n = 28). RESULTS: In index pregnancies with intravenous immunoglobulin treatment, fetal anemia developed on average 15 days later compared to previous pregnancies (8% less often <20 weeks' gestation). In pregnancies without intravenous immunoglobulin treatment anemia developed 9 days earlier compared to previous pregnancies (10% more <20 weeks), an adjusted 4-day between-group difference in favor of the immunoglobulin group (95% confidence interval, -10 to +18; P = .564). In the subcohort in which immunoglobulin treatment was started <13 weeks, anemia developed 25 days later and 31% less <20 weeks' gestation (54% compared to 23%) than in the previous pregnancy. Fetal hydrops occurred in 4% of immunoglobulin-treated pregnancies and in 24% of those without intravenous immunoglobulin treatment (odds ratio, 0.03; 95% confidence interval, 0-0.5; P = .011). Exchange transfusions were given to 9% of neonates born from pregnancies with and in 37% without immunoglobulin treatment (odds ratio, 0.1; 95% confidence interval, 0-0.5; P = .009). CONCLUSION: Intravenous immunoglobulin treatment in mothers pregnant with a fetus at risk for hemolytic disease seems to have a potential clinically relevant, beneficial effect on the course and severity of the disease. Confirmation in a multicenter randomized trial is needed.


Subject(s)
Anemia, Hemolytic/prevention & control , Erythroblastosis, Fetal/therapy , Immunoglobulins, Intravenous/therapeutic use , Immunologic Factors/therapeutic use , Adult , Anemia, Hemolytic/therapy , Blood Transfusion, Intrauterine , Disease Progression , Early Medical Intervention , Exchange Transfusion, Whole Blood/statistics & numerical data , Female , Fetal Diseases/therapy , Humans , Hydrops Fetalis/prevention & control , Infant, Newborn , Male , Odds Ratio , Pregnancy , Pregnancy Trimester, First , Pregnancy Trimester, Second , Retrospective Studies , Survival Rate , Time Factors
16.
J Pediatr Hematol Oncol ; 39(7): 524-527, 2017 10.
Article in English | MEDLINE | ID: mdl-28737602

ABSTRACT

Congenital thrombotic thrombocytopenia purpura (cTTP) is a very rare disorder worldwide. Standard treatment of recognized cases has been to administer fresh frozen plasma as the source of ADAMTS13, to replenish the absent ADAMTS13 enzyme. An alternative source, a plasma-derived factor VIII concentrate used for hemophilia A, and found to contain this enzyme, was reported to be effective in 1 patient in the United States. We now report details on a US cohort of 8 cTTP patients who have been successfully treated for varying periods with a marketed antihemophilic factor concentrate Koate-DVI. This biological product has been used successfully on demand in varying doses to treat acute exacerbations, as well as prophylactically (3 to 6 U ADAMTS13 every 3 to 21 d). Self-infused at home, in lieu of fresh frozen plasma therapy in the hospital setting, this product has effectively prevented episodes of thrombocytopenia, microangiopathic hemolytic anemia, and the concomitant organ damage in these patients. This specific virus inactivated product can be used to prevent further manifestations of this congenital enzyme deficiency.


Subject(s)
ADAMTS13 Protein/administration & dosage , Purpura, Thrombotic Thrombocytopenic/drug therapy , ADAMTS13 Protein/deficiency , Adolescent , Adult , Anemia, Hemolytic/prevention & control , Child , Cohort Studies , Factor VIII/administration & dosage , Factor VIII/chemistry , Humans , Thrombocytopenia/prevention & control , United States , Young Adult
18.
Elife ; 62017 02 04.
Article in English | MEDLINE | ID: mdl-28155819

ABSTRACT

Primaquine is the only drug available to prevent relapse in vivax malaria. The main adverse effect of primaquine is erythrocyte age and dose-dependent acute haemolytic anaemia in individuals with glucose-6-phosphate dehydrogenase deficiency (G6PDd). As testing for G6PDd is often unavailable, this limits the use of primaquine for radical cure. A compartmental model of the dynamics of red blood cell production and destruction was designed to characterise primaquine-induced haemolysis using a holistic Bayesian analysis of all published data and was used to predict a safer alternative to the currently recommended once weekly 0.75 mg/kg regimen for G6PDd. The model suggests that a step-wise increase in daily administered primaquine dose would be relatively safe in G6PDd. If this is confirmed, then were this regimen to be recommended for radical cure patients would not require testing for G6PDd in areas where G6PDd Viangchan or milder variants are prevalent.


Subject(s)
Anemia, Hemolytic/prevention & control , Antimalarials/adverse effects , Glucosephosphate Dehydrogenase Deficiency/drug therapy , Malaria, Vivax/drug therapy , Models, Statistical , Primaquine/adverse effects , Anemia, Hemolytic/chemically induced , Anemia, Hemolytic/diagnosis , Antimalarials/administration & dosage , Bayes Theorem , Cell Death/drug effects , Erythrocytes/drug effects , Female , Glucosephosphate Dehydrogenase Deficiency/complications , Glucosephosphate Dehydrogenase Deficiency/parasitology , Glucosephosphate Dehydrogenase Deficiency/pathology , Hemolysis/drug effects , Humans , Malaria, Vivax/complications , Malaria, Vivax/parasitology , Malaria, Vivax/pathology , Male , Plasmodium vivax/drug effects , Plasmodium vivax/growth & development , Primaquine/administration & dosage , Recurrence
20.
Turk J Gastroenterol ; 27(1): 55-61, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26674982

ABSTRACT

BACKGROUND/AIMS: Combination therapy with peginterferon (PEG-IFN) and ribavirin (RBV) has been recommended as a standard therapy for patients with chronic hepatitis C virus (HCV). Our aim was to evaluate the efficacy of eicosapentaenoic acid (EPA) against RBV-associated hemolytic anemia. MATERIALS AND METHODS: Two hundred and forty HCV patients included in the study were randomized to either the EPA group (n=120) or non-EPA group (n=120), and they received combination therapy with or without EPA. We compare changes in hemoglobin levels with RBV dose reduction rate in each group as well as treatment response. RESULTS: Of 120 patients randomized to receive combination therapy with EPA, 15/86 (17.5%) patients required RBV dose reduction, whereas 71/86 (82.5%) patients did not require RBV dose reduction; in the non-EPA group, 22/80 (27.5%) patients required RBV dose reduction and 58/80 (72.5%) patients did not require RBV dose reduction. There was no significant difference between the two groups in the rates of virologic response. CONCLUSION: EPA can decrease the rate of RBV dose reduction and RBV-induced hemolysis during the course of combination treatment. Further trials are required to investigate the role of EPA in the current regimens of HCV treatment that include ribavirin.


Subject(s)
Anemia, Hemolytic/chemically induced , Antiviral Agents/administration & dosage , Eicosapentaenoic Acid/administration & dosage , Hepatitis C, Chronic/drug therapy , Ribavirin/administration & dosage , Adult , Anemia, Hemolytic/prevention & control , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Hemoglobins/drug effects , Hemoglobins/metabolism , Hepacivirus/drug effects , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/virology , Humans , Interferon alpha-2 , Interferon-alpha/administration & dosage , Male , Middle Aged , Polyethylene Glycols/administration & dosage , Recombinant Proteins/administration & dosage , Ribavirin/adverse effects , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...